scholarly journals B cells lacking RelB are defective in proliferative responses, but undergo normal B cell maturation to Ig secretion and Ig class switching.

1996 ◽  
Vol 184 (4) ◽  
pp. 1537-1541 ◽  
Author(s):  
C M Snapper ◽  
F R Rosas ◽  
P Zelazowski ◽  
M A Moorman ◽  
M R Kehry ◽  
...  

A number of distinct functional abnormalities have been observed in B cells derived from p50/ NF-kappa B or c-rel knockout mice. RelB, another member of the NF-kappa B/Rel family of transcription factors, is expressed during the latter stages of B cell maturation and can bind to regulatory sites within the Ig heavy chain locus. Therefore, we tested the ability of B cells from relB knockout mice (relB-/-) to proliferate, undergo maturation to IgM secretion, and switch to the expression of downstream Ig isotypes in response to distinct activators including LPS, anti-CD40 mAb or CD40 ligand, and/or dextran anti-IgD antibodies in combination with various cytokines, including IL-4, IL-5, IFN-gamma, and TGF-beta. B cells lacking RelB showed up to 4-fold reductions in DNA synthesis in response to LPS, CD40, and membrane Ig-dependent activation relative to controls. However, relB-/- B cells were comparable to control B cells in their ability to undergo maturation to IgM secretion and switch to the expression of IgG3, IgG1, IgG2b, IgG2a, IgE, and/or IgA under all activation conditions tested. Thus, RelB, like c-Rel and p50/NF-kappa B, plays a role in B cell proliferation. However, in contrast to c-Rel and p50/ NF-kappa B, it is not critically involved in maturation to Ig secretion or expression of Ig isotypes.

Gut ◽  
2020 ◽  
Vol 69 (12) ◽  
pp. 2203-2213 ◽  
Author(s):  
Anton Lutckii ◽  
Benedikt Strunz ◽  
Anton Zhirkov ◽  
Olga Filipovich ◽  
Elena Rukoiatkina ◽  
...  

ObjectivesVertical transmission of hepatitis C virus (HCV) is rare compared with other chronic viral infections, despite that newborns have an immature, and possibly more susceptible, immune system. It further remains unclear to what extent prenatal and perinatal exposure to HCV affects immune system development in neonates.DesignTo address this, we studied B cells, innate immune cells and soluble factors in a cohort of 62 children that were either unexposed, exposed uninfected or infected with HCV. Forty of these infants were followed longitudinally from birth up until 18 months of age.ResultsAs expected, evidence for B cell maturation was observed with increased age in children, whereas few age-related changes were noticed among innate immune cells. HCV-infected children had a high frequency of HCV-specific IgG-secreting B cells. Such a response was also detected in some exposed but uninfected children but not in uninfected controls. Consistent with this, both HCV-exposed uninfected and HCV-infected infants had evidence of early B cell immune maturation with an increased proportion of IgA-positive plasma cells and upregulated CD40 expression. In contrast, actual HCV viraemia, but not mere exposure, led to alterations within myeloid immune cell populations, natural killer (NK) cells and a distinct soluble factor profile with increased levels of inflammatory cytokines and chemokines.ConclusionOur data reveal that exposure to, and infection with, HCV causes disparate effects on adaptive B cells and innate immune cell such as myeloid cells and NK cells in infants.


2000 ◽  
Vol 192 (1) ◽  
pp. 129-136 ◽  
Author(s):  
Jeffrey S. Thompson ◽  
Pascal Schneider ◽  
Susan L. Kalled ◽  
LiChun Wang ◽  
Eric A. Lefevre ◽  
...  

The tumor necrosis factor (TNF) family member B cell activating factor (BAFF) binds B cells and enhances B cell receptor–triggered proliferation. We find that B cell maturation antigen (BCMA), a predicted member of the TNF receptor family expressed primarily in mature B cells, is a receptor for BAFF. Although BCMA was previously localized to the Golgi apparatus, BCMA was found to be expressed on the surface of transfected cells and tonsillar B cells. A soluble form of BCMA, which inhibited the binding of BAFF to a B cell line, induced a dramatic decrease in the number of peripheral B cells when administered in vivo. Moreover, culturing splenic cells in the presence of BAFF increased survival of a percentage of the B cells. These results are consistent with a role for BAFF in maintaining homeostasis of the B cell population.


2006 ◽  
Vol 105 (1) ◽  
pp. 48-54 ◽  
Author(s):  
T NOJIMA ◽  
K HAYASHI ◽  
R GOITSUKA ◽  
K NAKAYAMA ◽  
K NAKAYAMA ◽  
...  

2006 ◽  
Vol 43 (9) ◽  
pp. 1316-1324 ◽  
Author(s):  
Ingela Wikström ◽  
Ingela Bergqvist ◽  
Dan Holmberg ◽  
Johan Forssell

2019 ◽  
Author(s):  
Samantha A. Swenson ◽  
Tyler J. Gilbreath ◽  
Heather Vahle ◽  
R. Willow Hynes-Smith ◽  
Jared H. Graham ◽  
...  

ABSTRACTCoordination of a number of molecular mechanisms including transcription, alternative splicing, and class switch recombination are required to facilitate development, activation, and survival of B cells. Disruption of these pathways can result in malignant transformation. Recently, next generation sequencing has identified a number of novel mutations in mantle cell lymphoma (MCL) patients including the ubiquitin E3 ligase UBR5. Approximately 18% of MCL patients were found to have mutations in UBR5 with the majority of mutations within the HECT domain of the protein which can accept and transfer ubiquitin molecules to the substrate. Determining if UBR5 controls the maturation of B cells is important to fully understand malignant transformation to MCL. To elucidate the role of UBR5 in B cell maturation and activation we generated a conditional mutant disrupting UBR5’s C-terminal HECT domain. Loss of the UBR5 HECT domain leads to a block in maturation of B cells in the spleen and up-regulation of proteins associated with mRNA splicing via the spliceosome. Our studies reveal a novel role of UBR5 in B cell maturation by regulating alternative splicing of key transcripts during B cell development and suggests UBR5 mutations may promote mantle cell lymphoma initiation.KEY POINTSUtilizing a novel mouse model mimicking MCL patient mutations, the loss of UBR5’s HECT domain causes alterations in B cell development.UBR5 mutations lead to stabilization of UBR5 and aberrant splicing.


2017 ◽  
Author(s):  
Joanne Dai ◽  
Micah A. Luftig

AbstractApoptosis is critical to B-cell maturation, but studies of apoptotic regulation in primary human B cells is lacking. Previously, we found that infecting human B cells with Epstein-Barr virus induces two different survival strategies (Priceet al., 2017). Here, we sought to better understand the mechanisms of apoptotic regulation in normal and activated B cells. Using intracellular BH3 profiling (iBH3), we defined the Bcl2-dependency of B-cell subsets from human peripheral blood and tonsillar lymphoid tissue as well as mitogen-activated B cells. We found that naïve and memory B cells were BCL-2 dependent, while germinal center B cells were MCL-1 dependent and plasma cells were BCL-XL dependent. Proliferating B cells activated by CpG or CD40L/IL-4 became more dependent upon MCL-1 and BCL-XL. As B-cell lymphomas often rely on survival mechanisms derived from normal and activated B cells, these findings offer new insight into potential therapeutic strategies for lymphomas.


Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 279-279
Author(s):  
Karen L. Bunting ◽  
David Soong ◽  
Yanwen Jiang ◽  
Brandon L. Swed ◽  
Katerina Hatzi ◽  
...  

Abstract Abstract 279 B cell affinity maturation is fundamental to the development of humoral immunity. To create a diverse antibody repertoire, B cells activated in the germinal centre (GC) must undergo a profound change in phenotype. This unique phenotypic change, which features simultaneous proliferation and somatic hypermutation and which can predispose to the development of lymphoma, requires radically altered gene expression programming in GC B cells. However, the way that this gene expression program is coordinated is unknown. Emerging evidence suggests that the higher-order organization of chromatin plays a role in the co-regulation of genes. We hypothesised that the three-dimensional organization of genes and chromosomes in the nucleus of B cells plays a key role in the epigenetic and transcriptional reprogramming that underlies acquisition of the GC B cell phenotype during B cell maturation. Using genome-wide mapping of chromatin interactions (Hi-C), combined with genome-wide profiles of gene expression (RNA-seq), histone modifications and transcription factor binding (ChIP-seq) in human naïve B (NB) and GC B cells, we have discovered that the three-dimensional structure of the genome undergoes widespread reorganization during B cell maturation to coordinate the GC transcriptional programme. Conformational maps of chromosome folding in these cells reveal a novel and profound loss of inter-arm interactions, reflecting lower chromosome compaction in GC B cells. Remarkably, we observed extensive differential partitioning of genes into NB- and GC B cell-specific compartments, and demonstrate for the first time that coordinated changes in histone modifications (H3K4Me2: P=3×10−35; H3K27Ac: P=3×10−33; Fisher's exact test) and transcription (P=1×10−9) required for cell type specification is mediated by the de novo formation of precisely delimited chromosome neighbourhoods. Most strikingly, we find that remodelling of the GC B cell genome involves the specific structural unlocking of genes that drive the GC transcriptional programme, such as AICDA, MTA3, and BCL6. Coordinate activation of these genes is mediated by the expansion of gene interaction neighbourhoods, increased promoter interactivity (P=3×10−35), engagement of long-range enhancer-promoter interactions (>2-fold increase), and the formation of gene body loops (P=3.18×10−15). Intriguingly, the master regulator of GC B cell differentiation, BCL6, shows a high propensity for all of these different types of interactions, suggesting that regulation of this gene in the context of chromatin is highly complex. Integration with genome-wide binding data for the structural organizing proteins, CTCF and cohesin, as well as the cell-specific factor, PU.1, supports a specific role for these proteins in the repositioning of activated promoters and enhancer regions during B cell maturation. This study shows for the first time that the architecture of the genome is critical for specification of cellular phenotype, and that epigenetic and transcriptional reprogramming in GC B cells is functionally linked to the structural reorganization of genes in the nucleus. Importantly, the higher-order organization of chromatin could represent a novel mechanism by which GC B cell gene expression is dysregulated in lymphoma. Disclosures: No relevant conflicts of interest to declare.


1974 ◽  
Vol 140 (3) ◽  
pp. 742-763 ◽  
Author(s):  
Jan Andersson ◽  
Joel Buxbaum ◽  
Ronald Citronbaum ◽  
Steven Douglas ◽  
Luciana Forni ◽  
...  

Five adjuvant induced BALB/c tumors producing IgM—McPc 1748, W 3469, TEPC 183, McPc 774, and Y 5781—were characterized morphologically by electron microscopy, analysis of the distribution of surface-bound and intracytoplasmic IgM using immunofluorescence, and by biochemical study of IgM synthesis, turnover, and secretion. The cells of different tumors appear to represent different stages in B-cell maturation when compared to normal, lipopolysaccharide-stimulated B cells. Thus, McPc 1748 tumor cells resemble 10–25-h stimulated normal B cells, 3469 cells resemble 20–35-h stimulated B cells, TEPC 183 cells resemble 45–65-h stimulated B cells, Y 5781 cells resemble 80–110-h stimulated B cells, and McPc 774 cells resemble 100–130-h stimulated B cells.


2017 ◽  
Vol 1 (27) ◽  
pp. 2729-2741 ◽  
Author(s):  
Julie Lang ◽  
Bicheng Zhang ◽  
Margot Kelly ◽  
Jacob N. Peterson ◽  
Jacob Barbee ◽  
...  

Key Points Expression of full-length hBAFF from cDNA in the endogenous murine locus does not improve maturation of human B cells in hu-mice. mBAFF is not limiting the maturation of human B cells in hu-mice.


Sign in / Sign up

Export Citation Format

Share Document